EP3679038A1 - Imidazoquinolines substituées - Google Patents

Imidazoquinolines substituées

Info

Publication number
EP3679038A1
EP3679038A1 EP18759137.5A EP18759137A EP3679038A1 EP 3679038 A1 EP3679038 A1 EP 3679038A1 EP 18759137 A EP18759137 A EP 18759137A EP 3679038 A1 EP3679038 A1 EP 3679038A1
Authority
EP
European Patent Office
Prior art keywords
alkyl
group
mmol
butyl
imidazo
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP18759137.5A
Other languages
German (de)
English (en)
Other versions
EP3679038B1 (fr
Inventor
Christophe Henry
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biontech SE
Original Assignee
Biontech SE
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Biontech SE filed Critical Biontech SE
Publication of EP3679038A1 publication Critical patent/EP3679038A1/fr
Application granted granted Critical
Publication of EP3679038B1 publication Critical patent/EP3679038B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to imidazoquinoline derivatives and to pharmaceutical compositions containing the imidazoquinoline derivatives.
  • the imidazoquinoline derivatives are useful as toll-like receptor agonists, in particular agonists of TLR7, and promote induction of certain cytokines.
  • TLR Toll-like receptors
  • TLR4 stimulated by E.coli lipopolysaccharide LPS
  • TLR3, 7, 8 and 9 located at endosomal membranes in specified immune cells.
  • TLR9 is activated by single-stranded DNA containing CpG subsequences
  • TLR7 and 8 are activated by single-stranded RNA
  • TLR3 is activated by double-stranded RNA.
  • TLR7 or TLR8 agonists have been identified. Those agonists can be grouped into purine-like molecules, such as 7-thia-8 -oxoguanosine (TOG, isatoribine) or the imidazoquinoline-based compounds like imiquimod. Imiquimod is so far the only approved definitive TLR7 agonist, marketed as 5% cream (by Aldara). It generates approx. 80% 5-year clearance of superficial basal cell carcinomas, which is the most frequent cancer worldwide. Imiquimod activates TLR7. The functional expression of TLR7 appears to be restricted to specific immune cells, i.e. in humans plasmacytoid dendritic cells, B-cells and probably eosinophils are known to be activated by TLR7 agonists.
  • TLR7 small-molecule
  • TLR7, 8 or 9 agonists are in clinical development for cancer mono- or combination therapies, or as vaccine adjuvant.
  • TLR agonist mediated immune activation is pleiotropic via specified immune cells (primarily dendritic cells and B-cells, subsequently other cells), which generates an innate and adaptive immune response. Moreover, not only one interferon is induced, but rather the many different isoforms altogether, and not only type I (alpha, beta), but also (indirectly) type II (gamma, NK cells). At least for local application, Aldara has delivered a remarkable proof-of-concept. This demonstrates that antigens are released by tumors, and that immune therapy can work for cancer indications in principle, and even in monotherapy.
  • TLR7 agonists For a systemic administration route, though, the clinical POC is pending for TLR7 agonists. For advanced cancers and systemic application (particularly s.c. or i.v. administration route) it appears to be clear that such TLR agonists might provide stronger, i.e. synergistic, efficacy in combination with other therapeutic interventions.
  • TLR toll-like receptor
  • TLR9 agonistic CpG- ODN like H2006 or HI 826 and TLR7 agonists like the guanosine derivative isatoribine or an Imiquimod derivative were tested in a murine Renca lung metastasis model. All tested molecules virtually completely suppressed the emergence of lung metastases with good tolerability. This provides a convincing rational for clinical development of such molecules for suppression of cancer metastasis and points to the possibility of systemic application of such drugs.
  • the SMOL type TLR7 agonists have the advantage of established and cost effective synthesis if compared to the nucleic acid type TLR9 agonists, and are well suited for topical application.
  • US-B-6,573,273 describes imidazoquinoline and tetrahydroimidazoquinoline compounds that contain urea, thiourea, acylurea, sulfonylurea or carbamate functionality. The compounds are said to be useful as immunomodulators.
  • US-B-6,677,349 describes imidazoquinoline and tetrahydroimidazoquinoline compounds that contain sulfonamide functionality at the 1 -position. The compounds are said to be useful as immunomodulators.
  • US-A-2003/0144283 and WO-A-00/76505 describe imidazoquinoline and tetrahydroimidazoquinoline compounds that contain amide functionality at the 1 - position. The compounds are said to be useful as immunomodulators.
  • WO- A-2005/051324 describes imidazoquinoline, pyridine and naphthyridine rind systems substituted in 1 -position with oxime or a special N -oxide functionality. The compounds are said to be useful as immunomodulators.
  • WO-A-2009/118296 describes imidazoquinoline compounds. The compounds are described as toll-like receptor agonist/TLR7 activators. Summary of the Invention:
  • the present invention provides imidazoquinoline-4-amine compounds with certain specific substituents, physiologically functional derivatives, solvates and salts thereof, as further described in the following.
  • Said compounds are agonists or activators for TLR7 and may serve as cytokine inducing compounds.
  • Said compounds have the general Formula (I):
  • Rl , R2, R3 and n are as defined below.
  • the present invention provides methods for the preparation of certain compounds of Formula (I), physiologically functional derivatives, solvates or salts thereof, as detailed further herein below.
  • the present invention provides methods for the treatment or prevention of certain medical conditions, the method comprising the administration of compounds of Formula (I), physiologically functional derivatives, solvates or salts thereof, to a subject in need thereof, as detailed further herein below.
  • the present invention provides the use of compounds of Formula (I), physiologically functional derivatives, solvates or salts thereof, in the manufacture of a medicament for the treatment or prevention of certain medical conditions, as detailed further herein below.
  • the present invention provides compounds of Formula (I), physiologically functional derivatives, solvates or salts thereof, for use as medicament, in particular for use in the treatment or prevention of certain medical conditions, as detailed further herein below.
  • the present invention provides pharmaceutical compositions comprising compounds of Formula (I), physiologically functional derivatives, solvates or salts thereof and one or more pharmaceutically acceptable excipients.
  • imidazoquinoline derivatives of Formula (I), physiologically functional derivatives, solvates or salts thereof, which are described in greater detail below, are particularly effective TLR7 agonists and have surprising and particularly advantageous properti es .
  • the present invention provides compounds of Formula (I):
  • Rl is selected from the group consisting of -H, G-6-alkyl, Ci-6-alkoxy, C1-3- alkoxy-Ci-3-alkyl, d-6-alkylthio, C 1.3-alkylthio-C 1 -3-alkyl, Ci-3-alkylamino- Ci-3-alkyl, 4- to 10-membered heterocycloalkyl, C3-io-cycloalkyl, Ce-io-aryl, C6-io-aryl-Ci -2 -alkyl and 5- to 10-membered heteroaryl,
  • -CO-R5, -CONH-R5, and -COO-R5 is selected from the group consisting of -CO-R5, -CONH-R5, and -COO-R5; is tetrahydropyran-4-yl, which is optionally substituted by one or more groups independently selected from the group consisting of Ci-4-alkyl, -OH, and halogen;
  • Ci-4-alkyl is each independently selected from the group consisting of H and Ci-4-alkyl; is an integer from 3 to 6;
  • Ci-e-alkyl is selected from the group consisting of -H, Ci-e-alkyl, Ci-6-alkoxy, C1-3- alkoxy-Ci-3-alkyl, Ci-6-alkylthio, Ci-3-alkylthio-Ci-3-alkyl, Ci-3-alkylamino- Ci-3-alkyl, Ce-io-aryl, 4- to 10-membered heterocycloalkyl, C 3 -io-cycloalkyl and 5- to 10-membered heteroaryl,
  • Rl is selected from the group consisting of Ci-6-alkyl, C 1 -3 -alkoxy-C 1 -3-alkyl, C 1 -3-alkylthio-C 1 -3-alkyl, and C1-3- alkylamino-C 1 -3-alkyl , more particularly Ci-e-alkyl, C 1 -3-alkoxy-C 1 -3-alkyl, and C - alkylamino-C I -3-alkyl, wherein said Ci-6-alkyl, Ci -3-alkoxy-C] -3-alkyl, C1 -3- alkylthio- Ci-3-alkyl, or Ci-3-alkylamino-Ci- 3 -alkyl is optionally substituted by one or more groups independently sel ected from the group consisting of -OH and halogen.
  • Rl is selected from the group consisting of ethyl, methyl, propyl, butyl, methoxyethyl, and ethylaminomethyl, each of which is optionally substituted by one or more groups independently selected from the group consisting of -OH and halogen, yet even more particularly each of which is unsubstituted.
  • Rl is selected from the group consisting of ethyl, propyl, butyl, methoxyethyl, and ethylaminomethyl, each of which is optionally substituted by one or more groups independently selected from the group consisting of -OH and halogen, yet even more particularly each of which is unsubstituted.
  • Rl is selected from the group consisting of ethyl, methoxyethyl, and ethylaminomethyl, each of which is optionally substituted by one or more groups independently selected from the group consisting of -OH and halogen, yet even more particularly each of which is unsubstituted.
  • Rl is unsubstituted methoxyethyl.
  • Rl is unsubstituted ethylaminomethyl .
  • R2 is selected from the group consisting of -CO-R5 and -CONH-R5, more particularly -CO-R5.
  • R3 is unsubstituted tetrahydropyran-4-yl.
  • R4 is each independently selected from the group consisting of H and methyl, more particularly H.
  • n 4.
  • R5 is selected from the group consisting of H, Ci-4-alkyl, Ci-3-alkoxy-Ci-3-alkyl, Ci -3-alkylamino-C i -3-alkyl, phenyl, 5- to 6-membered heterocycloalkyl, Cs-6-cycloalkyl and 5- to 6-membered heteroaryl, more particularly H, Ci-4-alkyl, C i -3-alkoxy-C i -3-alkyl, phenyl, and Cs-e-cycloalkyl, even more particularly H, O-4-alkyl, wherein said Ci-4-alkyl, C 1 -3-alkoxy-C 1 -3-alkyl, Ci-3-alkylarnino-Ci-3-alkyl, phenyl, 5- to 6-membered heterocycloalkyl, Cs-6- cycloalkyl and 5- to 6-membered heteroaryl is optionally substituted by one or more groups
  • R2 is -CO-R5, wherein R5 is selected from the group consisting of Ci-4-alkyl, C 1.3-alkoxy-C 1 -3-alkyl, and C1-3- alkylamino-C 1.3-alkyl, more particularly Ci-4-alkyl and Ci .3-alkoxy-C 1 -3-alkyl, more particularly R5 is Ci- -alkyl, wherein said Ci-4-alkyl, Ci-3-alkoxy-Ci-3-alkyl, and C1-3- alkylamino-Ci -3-alkyl is optionally substituted by one or more groups independently selected from the group consisting of Ci-2-alkyl, -OH, halogen, -CO-N(R4) 2 , -N(R4) 2 , -CO-R4, -COO-R4, -N 3 , -NO2, and -CN, more particularly Ci- 2 -alkyl, -OH, halogen,
  • R2 is CO-R5, wherein R5 is selected from the group consisting of Ci-4-alkyl, Ci-3-alkoxy-C
  • R2 is -CONH-R5, wherein R5 is selected from the group consisting of H, Ci-4-alkyl, C 1 -3-alkoxy-C 1 -3-alkyl , and C1-3- alkylamino-Ci-3-alkyl, more particularly H and Ci-4-alkyl, wherein said Ci-4-alkyl, Ci-3-alkoxy-C] -3-alkyl, and C 1.3-alkylamino-C 1-3-alkyl is optionally substituted by one or more groups independently selected from the group consisting of Ci-2-alkyl, -OH, halogen, -CO-N(R4) 2 , -N(R4) 2 , -CO-R4, -COO-R4, -N 3 , -N0 2 , and -CN, more particularly Ci.
  • R2 is -CONH-R5, wherein R5 is selected from the group consisting of H, Ci-4-alkyl, Ci-3-alkoxy-Ci-3- alkyl, and Ci-3-alkylamino-Ci-3-alkyl, more particularly H and Ci-4-alkyl, wherein said Ci-4-alkyl, Ci-3-alkoxy-Ci-3-alkyl, and C i .3-alkylamino-C 1-3-al kyl is unsubstituted.
  • R2 is -CONH 2 , -CO-Me, -COOMe, or -COOH, particularly -CONH2, -CO-Me, or -COOMe, more particularly -CONH2 or -CO-Me.
  • Rl is selected from the group consisting of Ci-6-alkyl, Ci-3-alkoxy-Cj -3 -alkyl, Ci-3-alkylthio-Ci -3-alkyl, and C 1 -3- alkylammo-Ci -3-alkyl, more particularly Ci-6-alkyl, Ci-3-alkoxy-Ci- 3 -alkyl, and C 1 -3- alkylamino-Ci -3-alkyl, wherein said O-6-alkyl, C 1 -3-alkoxy-C 1 -3-alkyl , Ci -3 - alkylthio- Ci -3-alkyl, or Ci- 3 -alkylamino-Ci- 3 -alkyl is optionally substituted by one or more groups independently selected from the group consisting of -OH and halogen; and R2 is -CO-R5, wherein R5 is selected from the group consisting of Ci- -alkyl, Ci
  • Ci-4-alkyl and C 1 -3- alkoxy-Ci-3-alkyl more particularly Ci-4-alkyl and C 1 -3- alkoxy-Ci-3-alkyl, more particularly R5 is Ci-4-alkyl, wherein said Ci-4-alkyl, C 1 -3- alkoxy-Ci -3-alkyl, and C t -3-alkyl amino-C 1 -3-alkyl is optionally substituted by one or more groups independently selected from the group consisting of G-2-alkyl, -OH, halogen, -CO-N(R4) 2 , -N(R4) 2 , -CO-R4, -COO-R4, -N 3 , -NO2, and -CN, more particularly Ci.
  • R2 is CO-R5, wherein R5 is selected from the group consisting of Ci-4-alkyl, C] -3-alkoxy-Ci-3-alkyl, and G-3-alkylamino-Ci -3-alkyl, more particularly Ci-4-alkyl and G -3-alkoxy-C 1 3- alkyl, more particularly R5 is G-4-alkyl, wherein said Ci-4-alkyl, C1.3-alkoxy-C1.3- alkyl, and C 1 -3-alkylamino-C 1 -3-alkyl is unsubstituted.
  • R3 is unsubstituted tetrahydropyran-4-yl and/or n is 4.
  • Rl is selected from the group consisting of G-6-alkyl, Ci-3-alkoxy-Ci-3-alkyl, C 1-3-alkylthio-Ci .3-alkyl, and C1-3- alkylamino-Ci-3-alkyl, more particularly G-6-alkyl, Ci -3-alkoxy-C 1 -3-alkyl, and C1-3- alkylamino-C 1 -3-alkyl, wherein said Ci-6-alkyl, C 1 -3-alkoxy-C 1 -3-alkyl, C1-3- alkylthio- Ci-3-alkyl, or Ci- 3 -alkylamino-Ci-3-alkyl is optionally substituted by one or more groups independently selected from the group consisting of -OH and halogen; and R2 is
  • R2 is -CONH-R5, wherein R5 is selected from the group consisting of H, Ci-4-alkyl, Ci- 3 -alkoxy-Ci-3-alkyl, and C 1 -3 -alkyl amino-C 1.3-alkyl , more particularly H and Ci-4-alkyl, wherein said Ci-4-alkyl, C 1 -3-alkoxy-C 1.3-alkyl , and C1-3- alkylamino-Ci- 3 -alkyl is unsubstituted.
  • R3 is unsubstituted tetrahydropyran-4-yl and/or n is 4.
  • tetrahydropyran-4-yl refers to a group of the below formula, wherein the interrupted bond specifies the point of attachment to the central moiety.
  • alkyl and the prefix “alk” are inclusive of both straight chain and branched chain groups and include the respective alkane, alkene and alkyne groups.
  • alkene and alkyne groups cannot consist only of a single carbon unit and such nonexistent groups are not comprised by the present invention; accordingly, and logically, terms such as Ci -x -alkyl (wherein x is an integer as specified in the respective context) include the respective Ci -x -alkanyl, C 2-x -alkenyl and C2-x-alkynyl.
  • Particular alkyl groups have a total of up to 5, particularly up to 4, more particularly up to 3 carbon atoms.
  • alkyl only refers to alkanyl groups (i.e., excluding alkenyl and alkynyl groups), in particular those alkanyl groups shown above (i.e., -CH 3 , -C 2 Hs, -Q5H7, etc.). All of the aforementioned alkyl groups, unless specified otherwise, are optionally substituted as detailed in the embodiments of the present invention, i.e. one or more hydrogen atoms are optionally replaced by a substituent as specified in said respective embodiment. Especially particularly, said alkyl groups are unsubstituted unless specified otherwise.
  • a particular form of an alkyl group is a haloalkyl group, which is an alkyl group as defined above, wherein one or more, particularly at least half, more particularly all of the hydrogen atoms on the hydrocarbon chain are replaced by halogen atoms.
  • the haloalkyl group is particularly selected from the group consisting of -C(R7)3, -CH 2 - C(R7) 3 , -C(R7) 2 -CH 3 , -C(R7) 2 -C(R7)3, -C(R7) 2 -CH(R7) 2 , -CH 2 -CH(R7) 2 , -CH(R7)- C(R7) 3 , -CH(R7)-CH 3 , and -C 2 H4-C(R7) 3 , more particularly -C(R7) 3 , wherein R7 represents halogen, particularly F. More particularly, haloalkyl is selected from the group consisting of -CF 3 , -CHF , -CH 2 CF 3 , and -CF2CI, even more particularly haloalkyl is -CF 3 .
  • alkynyl particularly refers to an alkyl group having at least two carbon atoms and including a carbon-carbon triple bond. Substituted alkynyl is as defined above.
  • alkenyl particularly refers to an alkyl group having at least two carbon atoms and including a carbon-carbon double bond.
  • a heteroaryl group particularly denotes an aromatic mono- or bicyclic hydrocarbon ring system wherein one or more carbon atoms are replaced by heteroatoms independently selected from the group consisting of O, N and S, wherein in the case of a monocyclic heteroaryl, said monocyclic heteroaryl may optionally be fused to a cycloalkyl or heterocycloalkyl ring, and wherein the total number of ring atoms in the heteroaryl group is five to ten, more particularly five or six.
  • the point of attachment of said heteroaryl group to the central moiety may be located on the mono- or bicyclic hydrocarbon ring system or on the optionally fused cycloalkyl or heterocycloalkyl ring.
  • heteroaryl group examples include thiadiazolc, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, isothiazol-3-yl, isothiazol-4-yl, isothiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl, 1,2,4-oxadiazol- 3-yl, 1 ,2,4-oxadiazol-5-yl, l,2,5-oxadiazol-3-yl, benzoxazol-2-yl, benzoxazol-4-yl, benzoxazol-5-yl, benzisoxazol-3 -yl, benzisoxazol-4-yl, benzisoxazol-5-yl, 1,2,5- oxadiazol-4-yl,
  • a cycloalkyl group particularly denotes a non-aromatic, mono- or bicyclic completely saturated or partially unsaturated hydrocarbon ring system, including bicyclic ring systems wherein one of the rings is a phenyl ring, such as 1 ,2,3,4-tetrahydronaphthalene. Said cycloalkyl is particularly monocyclic.
  • Said cycloalkyl is particularly completely saturated.
  • Said cycloalkyl comprises 3 to 10 carbon atoms, more particularly 5 to 7 carbon atoms.
  • said cycloalkyl is selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, 1-norbomyl, 2-norbornyl, 7-norbornyl, 1 - adamantyl, 2-adamantyl, 1 ,2-dihydronaphthyl, ,2,3 ,4-tetrahydronaphthyl, 2,3- dihydroindenyl, 1 ,6-dihydropentalenyl, 1 ,6a-dihydropentalenyl, yet even more particularly said cycloalkyl is selected from the group consisting of cyclopropyl, cyclopentyl, cyclohexyl and a
  • cycloalkyl groups are optionally substituted as detailed in the embodiments of the present invention, i.e. one or more hydrogen atoms are optionally replaced by a substituent as specified in said respective embodiment.
  • said cycloalkyl groups are unsubstituted unless specified otherwise.
  • a heterocycloalkyl group particularly denotes a non-aromatic mono- or bicyclic completely saturated or partially unsaturated hydrocarbon ring system, wherein one or more of the carbon atoms are replaced by a heteroatom independently selected from the group consisting of N, O, and S.
  • Said heterocycloalkyl does particularly not comprise any aromatic rings.
  • Said heterocycloalkyl is particularly monocyclic.
  • Said heterocycloalkyl is particularly completely saturated.
  • Said heterocycloalkyl particularly comprises a sum of 4 to 10 ring atoms, more particularly a sum of 5 to 10 ring atoms, even more particularly a sum of 5 to 7 ring atoms, yet even more particularly a sum of 5 or 6 ring atoms.
  • heterocycloalkyl is selected from the group consisting of morpholinyl, piperidinyl, dioxanyl, piperazinyl, thiomorpholinyl, piperidinyl, pyrrolidinyl, tetrahydrofuranyl, isoxazolidinyl, thiomorpholinyl, tetrahydrothioiuranyl and tetrahydropyranyl, more particularly selected from the group consisting of morpholinyl, piperidinyl, dioxanyl, piperazinyl, thiomorpholinyl, piperidinyl, and pyrrolidinyl.
  • heterocycloalkyl groups are optionally substituted as detailed in the embodiments of the present invention, i.e. one or more hydrogen atoms are optionally replaced by a substituent as specified in said respective embodiment.
  • said heterocycloalkyl groups are unsubstituted unless specified otherwise.
  • a halo or halogen group particularly denotes fluorine, chlorine, bromine or iodine, particularly chlorine or fluorine.
  • an alkoxy group denotes an O-alkyl group, wherein the alkyl group is as defined above.
  • the alkoxy group is particularly selected from the group consisting of methoxy, ethoxy and propoxy, more particularly methoxy. Said aforementioned alkoxy groups are optionally substituted with one or more halogen atoms, particularly with one or more fluorine atoms.
  • an alkylthio group denotes an -S-alkyl group, wherein the alkyl group is as defined above, particularly methylthio.
  • Said aforementioned alkylthio groups are optionally substituted with one or more halogen atoms, particularly with one or more fluorine atoms.
  • an alkoxyalkyl group denotes an alkyl group substituted with an O- alkyl group, wherein the alkyl groups are as defined above, particularly selected from the group consisting of methoxyethyl, ethoxymethyl, methoxymethyl, propoxymethyl and methoxypropyl, more particularly methoxyethyl.
  • Said aforementioned alkoxyalkyl groups are optionally substituted with one or more halogen atoms, particularly with one or more fluorine atoms.
  • an alkylthioalkyl group denotes an alkyl group substituted with an S- alkyl group, wherein the alkyl groups are as defined above, particularly selected from the group consisting of methylthioethyl, ethylthiomethyl, methylthiomethyl, propylthiomethyl and methylthiopropyl, more particularly methylthioethyl.
  • Said aforementioned alkylthioalkyl groups are optionally substituted with one or more halogen atoms, particularly with one or more fluorine atoms.
  • an alkylaminoalkyl group denotes an alkyl group linked to an NH- alkyl group or N-dialkyl group, wherein the alkyl groups are as defined above, particularly selected from the group consisting of methylaminoethyl , ethylaminomethyl, methylaminomethyl, propylaminomethyl and methylaminopropyl, more particularly ethylaminomethyl.
  • Said aforementioned alkylaminoalkyl groups are optionally substituted with one or more halogen atoms, particularly with one or more fluorine atoms.
  • an aryl group particularly denotes an aromatic mono- or bicyclic hydrocarbon ring system, wherein the total number of ring atoms in the aryl group is six to ten, particularly six.
  • Examples of the aryl group are phenyl and naphthyl, more particularly phenyl. All of the aforementioned aryl groups, unless specified otherwise, are optionally substituted as detailed in the embodiments of the present invention, i.e. one or more hydrogen atoms are optionally replaced by a substituent as specified in said respective embodiment. Especially particularly, said aryl groups are unsubstituted unless specified otherwise.
  • arylalkyl group also commonly known as aralkyi group particularly denotes a linear or branched alkyl as defined herein substituted with an aryl group as defined herein.
  • exemplary arylalkyl groups include styryl, benzyl, phenylethyl, l-(naphthalen- 2-yl)ethyl, particularly the arylalkyl group is styryl or benzyl, more particularly benzyl.
  • Said aforementioned arylalkyl group is optionally substituted, particularly at its aryl part, as defined above for the aryl group.
  • alkyl aryl, arylalkyl, “heterocycloalkyl”, “cycloalkyl”, “heteroaryl”, “alkoxy”, “alkylthio”, “alkoxyalkyl”, “alkylthioalkyl”, “alkylaminoalkyl”, and the like, also refer, insofar as this is applicable, to specific members of said groups as specified in the embodiments of the present invention.
  • alkyl also refers, insofar as this is applicable, to members of said group, such as “Ci-6-alkyl", “Ci-4-alkyl", “Ci -2 -alkyl", methyl, ethyl, and the like.
  • a nitrogen heteroatom (N) as defined herein, e.g. in the context of "heteroaryl”, “heterocycloalkyl” and “heterocycle”, may include the N-oxide, in particular where chemically feasible from the viewpoint of stability and/or chemical valence rules.
  • a sulfur heteroatom (S) as defined herein, e.g. in the context of “heteroaryl”, “heterocycloalkyl” and “heterocycle”, may include the sulfur oxide and/or the sulfur dioxide, in particular where chemically feasible from the viewpoint of stability and/or chemical valence rules.
  • substituted with or “substituted by” means that one or more hydrogen atoms connected to a carbon atom or heteroatom of a chemical group or entity are exchanged with a substituent group, respectively; e.g. substituted aryl comprises 4-hydroxyphenyl, wherein the H-atom in the 4-position of the phenyl group is exchanged with a hydroxyl group.
  • Said hydrogen atom(s) to be replaced may be attached to a carbon atom or heteroatom, and may be expressly shown in a specific formula, such as for example in an -NH- group, or may not expressly be shown but intrinsically be present, such as for example in the typical "chain” notation which is commonly used to symbolize e.g.
  • substituent groups may be selected from the group consisting of Ci-4-alkyl, -OH, halogen, -CO-N( i) 2 , -N(Ri) 2 , -CO-Ri, -COO-Ri, -N., -N0 2 , and -CN, wherein Ri is each independently selected from the group consisting of H and C !-4 -alkyl.
  • references to the compounds according to the present invention include the physiologically functional derivatives, solvates or salts thereof as described herein, as well as to salts of said physiologically functional derivatives, solvates of salts and physiologically functional derivatives, and optionally solvates of salts of physiologically functional derivatives.
  • the term "physiologically functional derivative" of a compound according to the present invention is for instance a prodrug of said compound, wherein at least one of the following groups are derivatized as specified in the following:
  • a carboxylic acid (-COOH) group is derivatized into an ester (having e.g., the formula -COOR8, wherein R8 is selected from the group consisting of -H, alkyl (such as Ci-6- alkyl), alkoxy (such as Ci-6-alkoxy), alkoxyalkyl (such as Ci-3-alkoxy-Ci-3-alkyl), alkylthio (such as C]-6-alkylthio), alklylthioalkyl (such as Ci-3-alkylthio-Ci-3-alkyl), alkylaminoalkyl (such as Ci-3-alkylamino-Ci-3-alkyl), aryl (such as Ci-io-aryl), heterocycloalkyl (such as
  • the compounds according to the present invention are to be understood to comprise all tautomeric forms thereof, even if not expressly shown in the formulae described herein, including Formula (I).
  • the compounds of Formula (I) as defined herein are to be understood to encompass, where applicable, all stereoisomers of said compounds, unless specified otherwise.
  • stereoisomer refers to a compound with at least one stereogenic centre, which may be R- or S-configured, as defined by the according IUPAC rules, and encompasses enantiomers and diastereomers as commonly understood by the skilled person. It has to be understood, that in compounds with more than one stereogenic centre, each of the individual stereogenic centres may independently from each other be R- or S-configured.
  • the term “stereoisomer' * as used herein also refers to salts of the compounds herein described with optically active acids or bases.
  • the invention further includes all mixtures of the stereoisomers mentioned above independent of the ratio, including the racemates.
  • the salts of the compounds according to the present invention are particularly pharmaceutically acceptable salts of the compounds according to the present invention.
  • Pharmaceutically acceptable salts are such salts which are usually considered by the skilled person to be suitable for medical applications, e.g. because they are not harmful to subjects which may be treated with said salts, or which give rise to side effects which are tolerable within the respective treatment.
  • said pharmaceutically acceptable salts are such salts which are considered as acceptable by the regulatory authorities, such as the US Food and Drug Administration (FDA), the European Medicines Agency (EMA), or the Japanese Ministry of Health, Labor and Welfare Pharmaceuticals and Medical Devices Agency (PMDA).
  • the present invention in principle also encompasses salts of the compounds according to the present invention which are as such not pharmaceutically acceptable, e.g. as intermediates in the production of the compounds according to the present invention or physiologically functional derivatives thereof, or as intermediates in the production pharmaceutically acceptable salts of the compounds according to the present invention or physiologically functional derivatives thereof.
  • Said salts include water-insoluble and, particularly, water-soluble salts.
  • a certain compound according to the present invention or physiologically functional derivative thereof can form a salt, i.e. whether said compound according to the present invention or physiologically functional derivative thereof has a group which may carry a charge, such as e.g. an amino group, a carboxylic acid group, etc.
  • Exemplary salts of the compounds of the present invention are acid addition salts or salts with bases, particularly pharmaceutically acceptable inorganic and organic acids and bases customarily used in pharmacy, which are either water insoluble or, particularly, water-soluble acid addition salts. Salts with bases may - depending on the substituents of the compounds of the present invention - also be suitable. Acid addition salts may, for example, be formed by mixing a solution of a compound of the present invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
  • a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
  • pharmaceutically acceptable base addition salts may include alkali metal salts (e.g., sodium or potassium salts); alkaline earth metal salts (e.g., calcium or magnesium salts); and salts formed with suitable organic ligands (e.g., ammonium, quaternary ammonium and amine cations formed using counteranions such as halide, hydroxide, carboxylase, sulfate, phosphate, nitrate, alkyl sulfonate and aryl sulfonate).
  • suitable organic ligands e.g., ammonium, quaternary ammonium and amine cations formed using counteranions such as halide, hydroxide, carboxylase, sulfate, phosphate, nitrate, alkyl sulfonate and aryl sulfonate.
  • Illustrative examples of pharmaceutically acceptable salts include, but are not limited to, acetate, adipate, alginate, arginate, ascorbate, aspartate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, butyrate, calcium edetate, camphorate, camphorsulfonate, camsylate, carbonate, chloride, citrate, digluconate, dihydrochloride, dodecylsulfate, edetate, edisylate, ethanesulfonate, formate, fumarate, galactate, galacturonate, gluconate, glutamate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hexylresorcinate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, hydroxy
  • Salts which are not pharmaceutically acceptable and which can be obtained, for example, as process products during the preparation of the compounds according to the invention on an industrial scale, are also encompassed by the present invention and, if desired, may be converted into pharmaceutically acceptable salts by processes known to the person skilled in the art.
  • the compounds of the invention as well as their salts may contain, e.g. when isolated in crystalline form, varying amounts of solvents. Included within the scope of the invention are therefore solvates and in particular hydrates of the compounds of the present invention as well as solvates and in particular hydrates of the salts and/or physiologically functional derivatives of the compounds of the present invention. More particularly the invention encompasses hydrates of the compounds, salts and/or physiologically functional derivatives according to the present invention, comprising one, two or one half water molecule, with respect to their stoichiometry.
  • room temperature rt or “r.t.” relates to a temperature of from 20 to 25 °C, particularly about 22°C, unless specified otherwise.
  • stable specifies a compound in which the chemical structure is not altered when the compound is stored at a temperature from about -80 °C to about +40 °C, particularly from about -80 °C to +25 °C in the absence of light, moisture or other chemically reactive conditions for at least one week, particularly at least one month, more particularly at least six months, even more particularly, at least one year, and/or a compound which under IUPAC standard conditions and in the absence of light, moisture or other chemically reactive conditions maintains its structural integrity long enough to be useful for therapeutic or prophylactic administration to a patient, i.e.
  • a compound, in particular a compound of Formula (I), is selective for a predetermined target (in particular TLR7) if it is capable of binding to and exerting activity (in particular agonist activity) towards said predetermined target while it is not capable of binding to and exerting agonist activity (in particular agonist and antagonist activity) towards other targets, i.e. exerts no significant agonist activity for other targets in standard assays.
  • a compound of Formula (I) is selective for TLR7 if it is capable of exerting agonist activity towards TLR7 but is not (substantially) capable of exerting agonist activity towards other targets, in particular TLR8.
  • a compound, in particular a compound of Formula (I), is selective for TLR7 if the agonist activity for such other targets (in particular TLR8) does not significantly exceed the agonist activity for TLR-unrelated proteins such as LDL receptor, insulin receptor or transferrin receptor or any other specified polypeptide.
  • a compound, in particular a compound of Formula (I), is selective for a predetermined target (in particular TLR7) if its agonist activity (EC50) for said target is at least 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 15-fold, 20-fold, 25-fold, 30-fold, 35- fold, 40-fold, 45-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold or 10 3 -fold lower than the its agonist activity for a target for which it is not selective (in particular TLR8).
  • EC50 agonist activity
  • the ECso for a target for which the compound it is not selective would be at least 2 ⁇ , 3 ⁇ , 4 ⁇ , 5 ⁇ , 10 ⁇ , 15 ⁇ , 20 ⁇ , 25 ⁇ , 30 ⁇ , 35 ⁇ , 40 ⁇ , 45 ⁇ , 50 ⁇ , 60 ⁇ , 70 ⁇ , 80 ⁇ , 90 ⁇ , 100 ⁇ or 1 mM. It is to be understood that the invention covers all combinations of substituent groups referred to hereinabove. In particular, the invention covers all combinations of particular groups described hereinabove.
  • E isomers and Z isomers Compounds of the invention and salts thereof containing a double bond may exist as E isomers and Z isomers. Both said isomers are included in the invention.
  • the Z isomer is the geometric isomer in which the carbon atoms connected by the double bond each have the two highest ranking groups on the same side of the double bond.
  • the E isomer is the geometric isomer in which the carbon atoms connected by the double bond each have the two highest ranking groups on opposite sides of the double bond.
  • the term “treatment” includes complete or partial healing of a disease, prevention of a disease, alleviation of a disease or stop of progression of a given disease.
  • the terms “medical condition”, “disease”, and “disorder” are used herein interchangeably and refer to any pathological state, including proliferative diseases such as cancer, in particular those pathological states (including cancer forms) described herein.
  • a disease is characterized that it can be treated by agonizing TLR7.
  • a "proliferative disease” includes a disease characterized by aberrantly regulated cellular growth, proliferation, differentiation, adhesion, and/or migration.
  • a particular example of proliferative diseases is cancer.
  • cancer cell is meant an abnormal cell that grows by a rapid, uncontrolled cellular proliferation and continues to grow after the stimuli that initiated the new growth cease.
  • the term “medicament” includes the compounds of Formula (I) as described herein, pharmaceutically acceptable salts or physiologically functional derivatives thereof, which are to be administered to a subject in pure form, as well as compositions comprising at least one compound according to the present invention, a pharmaceutically acceptable salt or physiologically functional derivative thereof, which is suitable for administration to a subject.
  • the compounds according to the present invention and their pharmaceutically acceptable salts and physiologically functional derivatives can be administered to animals, particularly to mammals, and in particular to humans as therapeutics per se, as mixtures with one another or particularly in the form of pharmaceutical preparations or compositions which allow enteral (e.g. oral) or parenteral administration and which comprise as active constituent a therapeutically effective amount of at least one compound according to the present invention, or a salt or physiologically functional derivative thereof, in addition to e.g.
  • compositions, medical uses and methods of treatment according to the present invention may comprise more than one compound according to the present invention.
  • compositions comprising a compound according to the present invention, or a pharmaceutically acceptable salt or physiologically functional derivative may optionally comprise one or more further therapeutically active substances which are not compounds of Formula (I) according to the present invention.
  • therapeutically active substance specifies a substance which upon administration can induce a medical effect in a subject.
  • Said medical effect may include the medical effect described herein for the compounds of Formula (I) of the present invention, but may also, in the case of therapeutically active substances which are to be co-administered with the compounds according to the present invention, include other medical substances, such as for example but not exclusively irinotecan, oxaliplatin, gemcitabine, capecitabine, 5-fluorouracil, cetuximab (Erbitux), panitumumab (Vectibix), bevacizumab (Avastin), vincristine, vinblastine, vinorelbine, vindesine, taxol, amsacrine, etoposide, etoposide phosphate, tenyposide, actinomycin, anthracyclines, doxorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin, mitomycin, mechlorethamine, cyclophosphamide, chloram
  • pharmaceutically acceptable is well known to the skilled person and particularly means that the respective entity is not harmful to the subject to which the entity or the composition comprising the entity is administered, that said entity is stable and that said entity is chemically compatible (i.e. non-reactive) with other ingredients of the respective pharmaceutical composition.
  • Medicaments and pharmaceutical compositions according to the present invention comprising at least one compound according to the present invention or a pharmaceutically acceptable salt or a physiologically functional derivative thereof include those suitable for oral, rectal, bronchial, nasal, topical, buccal, sub-lingual, vaginal or parenteral (including transdermal, intracutaneous, subcutaneous, intramuscular, intrapulmonary, intravascular, intracranial, intraperitoneal, intravenous, intraarterial, intracerebral, intraocular, intrastemal, intracoronary, transurethral, injection or infusion) administration, or those in a form suitable for administration by inhalation or insufflation, including powders and liquid aerosol administration, or by controlled release (e.g.
  • sustained release, pH-controlled release, delayed release, repeat action release, prolonged release, extended release) systems include semipermeable matrices of solid hydrophobic polymers containing the compound of the invention, which matrices may be in form of shaped articles, e.g. films or microcapsules or colloidal drug carriers e.g. polymeric nanoparticles, or controlled release solid dosage forms, e.g. core tablets or multi-layer tablets.
  • a particular route of administration in the present invention is intravenous administration.
  • the compounds according to the present invention may particularly be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative.
  • the compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulation agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for re- constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • any of the other conventional dosage forms may also be used, such as tablets, lozenges, parenteral formulations, syrups, creams, ointments, aerosol formulations, transdermal patches, transmucosal patches and the like.
  • the pharmaceutical compositions according to the present invention can be formulated, for example, into tablets, coated tablets (dragees), pills, cachets, capsules (caplets), granules, powders, suppositories, solutions (e.g. sterile solutions), emulsions, suspensions, ointments, creams, lotions, pastes, oils, gels, sprays and patches (e.g. transdermal therapeutic systems).
  • the pharmaceutical compositions can be prepared as, e.g. liposome delivery systems, systems in which the active compound is coupled to monoclonal antibodies and systems in which the active compound is coupled to polymers (e.g. soluble or biodegradable polymers).
  • Tablets, coated tablets (dragees), pills, cachets, capsules (caplets), granules, solutions, emulsions and suspensions are, e.g. suitable for oral administration.
  • said formulations can be adapted so as to represent, for example, an enteric form, an immediate release form, a delayed release form, a repeated dose release form, a prolonged release form or a sustained release form.
  • Said forms can be obtained, for example, by coating tablets, by dividing tablets into several compartments separated by layers disintegrating under different conditions (e.g. pH conditions) or by coupling the active compound to a biodegradable polymer.
  • the aerosol is a liquid-gaseous dispersion, a solid-gaseous dispersion or a mixed liquid/solid-gaseous dispersion.
  • the particle size of the aerosol particles is particularly less than 100 ⁇ , more particularly it is in the range of from 0.5 to 10 ⁇ , even more in particular in the range of from 2 to 6 ⁇ (ID50 value, measured by laser diffraction).
  • the aerosol may be generated by means of aerosol-producing devices such as dry powder inhalers (DPIs), pressurized metered dose inhalers (PMDIs) and nebulizers.
  • the aerosol-producing device can contain the active compound in form of a powder, a solution or a dispersion.
  • the powder may contain, for example, one or more of the following auxiliaries: carriers, stabilizers and fillers.
  • the solution may contain in addition to the solvent, for example, one or more of the following auxiliaries: propellants, solubilizers (co-solvents), surfactants, stabilizers, buffers, tonicity adjusting agents, preservatives and flavorings.
  • the dispersion may contain in addition to the dispersant, for example, one or more of the following auxiliaries: propellants, surfactants, stabilizers, buffers, preservatives and flavorings.
  • auxiliaries include, but are not limited to, saccharides, e.g. lactose and glucose.
  • propellants include, but are not limited to, fluorohydrocarbons, e.g.
  • aerosol-producing devices which may be used for inhaled administration include, but are not limited to, Cyclohaler®, Diskhaler®, Rotadisk®, Turbohaler®, Autohaler®, Turbohaler®, Novolizer®, Easyhaler®, Aerolizer®, Jethaler®, Diskus®, Ultrahaler® and Mystic® inhalers.
  • the aerosol-producing devices may be combined with spacers or expanders, e.g. Aerochamber®, Nebulator®, Volumatic® and Rondo®, for improving inhalation efficiency.
  • Pharmaceutically acceptable carriers used in the preparation of a pharmaceutical composition or medicament comprising a compound according to the present invention, a pharmaceutically acceptable salt or physiologically functional derivative thereof can be either solid or liquid.
  • Solid form pharmaceutical compositions comprising a compound according to the present invention, a pharmaceutically acceptable salt or physiologically functional derivative thereof include powders, tablets, pills, capsules, sachets, suppositories, and dispersible granules.
  • a solid carrier may comprise one or more components, which may also act as diluents, flavouring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • the carrier is a finely divided solid, which is in a mixture with the finely divided active component.
  • the active component is mixed with the carrier having the necessary binding capacity in suitable proportions and compacted in the shape and size desired.
  • the tableting mixture can be granulated, sieved and compressed or direct compressed.
  • Suitable carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatine, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.
  • preparation is intended to include the formulation of the active compound with encapsulating material as carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it.
  • sachets and lozenges are included. Tablets, powders, capsules, pills, sachets, and lozenges can be used as solid forms suitable for oral administration.
  • a low melting wax such as a mixture of fatty acid glyceride or cocoa butter
  • the active component is dispersed homogeneously therein, as by stirring.
  • the molten homogenous mixture is then poured into conveniently sized moulds, allowed to cool, and thereby to solidify.
  • Compositions suitable for vaginal administration may be presented as peccaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • Liquid preparations include solutions, suspensions, and emulsions, for example, water or water-propylene glycol solutions.
  • parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution.
  • Aqueous solutions suitable for oral administration can be prepared by dissolving the active component in water and adding for example suitable colorants, flavours, stabilizing and thickening agents, as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well-known suspending agents.
  • viscous material such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well-known suspending agents.
  • solid form preparations which are intended to be converted, shortly before administration, to liquid form preparations for oral administration.
  • Such liquid forms include solutions, suspensions, and emulsions.
  • These preparations may contain, in addition to the active component, for example colorants, flavours, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.
  • the medicament is applied topically, e.g. in the form of transdermal therapeutic systems (e.g. patches) or topical formulations (e.g. liposomes, cremes, ointment, lotion, gels, dispersion, suspension, spray, solution, foam, powder).
  • transdermal therapeutic systems e.g. patches
  • topical formulations e.g. liposomes, cremes, ointment, lotion, gels, dispersion, suspension, spray, solution, foam, powder.
  • the medicament may comprise carrier materials or excipients, including but not limited to a lipophilic phase (as for example Vaseline, paraffines, triglycerides, waxes, polyalkylsiloxanes), oils (olive oil, peanut oil, castor oil, triglyceride oil), emulsifier (as for example lecithin, phosphatidylglyceroles, alkyl alcohols, sodium lauryl sulfate, polysorbats, Cholesterol, sorbitan fatty acid ester, polyoxyethylene fatty acid glycerol and -ester, poloxamers), preservatives (for instance benzalkonium chloride, chlorobutanol, parabene or thiomersal), flavouring agents, buffer substances (for example salts of acetic acid, citric acid, boric acid, phosphoric acid, tartaric acid, trometamole or trolamine), solvents (for instance polyethylengly
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or colouring agents.
  • Compositions suitable for topical administration in the mouth include lozenges comprising the active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatine and glycerine or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Solutions or suspensions may be applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray.
  • the compositions may be provided in single or multi-dose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension. In the case of a spray, this may be achieved for example by means of a metering atomizing spray pump.
  • Administration to the respiratory tract may also be achieved by means of an aerosol formulation in which the active ingredient is provided in a pressurized pack with a suitable propellant such as a chlorofluorocarbon (CFC), for example dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, carbon dioxide, or other suitable gas.
  • a suitable propellant such as a chlorofluorocarbon (CFC), for example dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, carbon dioxide, or other suitable gas.
  • CFC chlorofluorocarbon
  • the aerosol may conveniently also contain a surfactant such as lecithin.
  • the dose of drug may be controlled by provision of a metered valve.
  • the medicament may be provided in the form of a dry powder, for example a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • a powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • PVP polyvinylpyrrolidone
  • the powder carrier will form a gel in the nasal cavity.
  • the powder composition may be presented in unit dose form, for example in capsules or cartridges of, e.g., gelatine, or blister packs from which the powder may be administered by means of an inhaler.
  • compositions for administration to the respiratory tract including intranasal compositions
  • the compound will generally have a small particle size for example of the order of 5 microns or less. Such a particle size may be obtained by means known in the art, for example by micronization.
  • compositions adapted to give sustained release of the active ingredient may be employed.
  • the pharmaceutical preparations are particularly in unit dosage forms.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packaged tablets, capsules, and powders in vials or ampoules.
  • the unit dosage form can be a capsule, tablet, sachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form. Tablets or capsules for oral administration and liquids for intravenous administration and continuous infusion are particular compositions.
  • the compounds of the present invention may be used in combination with radiation therapy, or in combination with radiation therapy and other active compounds, already known for the treatment of the medical conditions disclosed herein, whereby a favourable additive or amplifying effect is noticed.
  • pharmaceutically inert inorganic or organic excipients can be used.
  • pills tablets, coated tablets and hard gelatine capsules, for example, lactose, cornstarch or derivatives thereof, talc, stearic acid or its salts, etc.
  • Excipients for soft gelatine capsules and suppositories are, for example, fats, waxes, semi-solid and liquid polyols, natural or hardened oils etc.
  • Suitable excipients for the production of solutions and syrups are, for example, water, sucrose, invert sugar, glucose, polyols etc.
  • Suitable excipients for the production of injection solutions are, for example, water, alcohols, glycerol, polyols or vegetable oils.
  • terapéuticaally effective amount means an amount of the compound sufficient to induce a therapeutic effect, such as activation of TLR7. This may cause cytokine induction, antitumor activity and/or antiviral activity.
  • amount of active compound used in a pharmaceutical composition of the invention will vary according to factors known to those skilled in the art, such as the physical and chemical nature of the compound as well as the nature of the carrier and the intended dosing regimen, it is anticipated that the compositions of the invention will contain sufficient active ingredient to provide a suitable dose to a subject. Said dose can vary within wide limits and is to be suited to the individual conditions in each individual case. For the medical applications of the present invention, the appropriate dosage will vary depending on the mode of administration, the particular condition to be treated and the effect desired.
  • dosage rates of the compound of the present invention of about 1 ng kg to 100 mg kg subject body weight, particularly 100 ng/kg to 10 mg/kg, more particularly 1 ⁇ g/ g to 1 mg kg.
  • Doses may be conveniently administered once in two weeks, once or several times per week or up to 2 to 4 times a day in divided doses or in sustained release form.
  • the compounds of the present application are selective agonists for TLR7 (especially over TLR8).
  • the inventors of the present application have found that the double substitution of the amino group NR2R3 of Formula (I) is associated with the TLR7 selectivity, whereas a mono-substitution of the amino group NR2R3 of Formula (I) (i.e., where R2 is H) leads to compounds which are selective for TLR8.
  • the compounds of the present application are suitable for the treatment of a medical condition, disease or disorder which can be treated by agonizing TLR7.
  • the compounds of the present invention are preferably suitable for the treatment of viral disorders and proliferative diseases, in particular hyperproliferative diseases, such as benign and malignant forms of neoplasia, including cancer.
  • Exemplary types of cancer in the context of the present invention are hepatocarcinoma, adrenocortical carcinoma, AIDS-related cancers including AIDS-related lymphoma, anal cancer, basal cell carcinoma, bile duct cancer, bone cancer, brain tumors including brain stem glioma, cerebellar astrocytoma, cerebral astrocytoma, malignant glioma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal tumors, visual pathway and hypothalamic glioma, breast cancer, bronchial adenomas/car cinoids, Burkitt's lymphoma, gastrointestinal carcinoma, carcinoma of unknown primary site, central nervous system lymphoma, cervical cancer, chronic myeloproliferative disorders, colon cancer, colorectal cancer, stomach cancer, cutaneous T-cell lymphoma, endometrial cancer, ependymoma, esophageal
  • the compounds of the present invention may be used in the treatment of the following cancer types: Prostate, bladder, kidney (i.e. renal), muscle, ovary, skin, stomach, pancreas, breast, cervix, colon, liver, connective tissue, placenta, bone, brain, uterus, salivary gland, or testes.
  • Illustrative cancers include, but are not limited to cancer of the breast, bladder, bone, brain, central and peripheral nervous system, colon, endocrine glands, esophagus, endometrium, germ cells, head and neck, kidney, liver, lung, larynx and hypopharynx, mesothelioma, sarcoma, ovary, pancreas, prostate, rectum, renal, small intestine, soft tissue, testis, stomach, skin, ureter, vagina and vulva; inherited cancers, retinomblastoma and Wilms' tumor; leukemia, lymphoma, non-Hodgkin's disease, chronic and acute myeloid leukaemia, acute lymphoblastic leukemia, Hodgkin's disease, multiple myeloma and T-cell lymphoma; myelodysplastic syndrome, plasma cell neoplasia, paraneoplastic syndromes, cancers of unknown primary site and AIDS- related malign
  • TLR7 agonists would be used to treat cancers of the skin, breast, colon, stomach, pancreas or kidney. Sensitivity of a given cancer to activation of TLR7 can be assessed by, but not limited to measurement of a decrease in primary or metastatic tumor load (minor, partial or complete regression), alterations in the hemogram, altered hormone or cytokine concentrations in the blood, inhibition of further increase of tumor load, stabilization of the disease in the patient, assessment of biomarkers or surrogate markers relevant for the disease, prolonged overall survival of a patient, prolonged time to disease progression of a patient, prolonged progression-free survival of a patient, prolonged disease-free survival of a patient, improved quality of life of a patient, or modulation of the co-morbidity of the disease (for example, but not limited to pain, cachexia, mobilization, hospitalization, altered hemogram, weight loss, wound healing, fever).
  • the co-morbidity of the disease for example, but not limited to pain, cachexia, mobilization, hospitalization, altered hemogram, weight loss
  • compositions according to the present invention may comprise one or more, particularly one or two, more particularly one of the compounds according to the present invention.
  • the medical applications of the present invention may involve one or more, particularly one or two, more particularly one of the compounds according to the present invention
  • compositions comprising the active compound and at least one auxiliary can be manufactured in a manner known to a person skilled in the art, e.g. by dissolving, mixing, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • suitable pharmaceutical formulations are, for example, ointments, creams, lotions, pastes, gels, powders, solutions, emulsions, suspensions, oils, sprays and patches (e.g. transdermal therapeutic systems).
  • parenteral modes of administration such as, for example, intravenous, intraarterial, intramuscular, subcutaneous, intracutaneous, intraperitoneal and intrasternal administration, particularly solutions (e.g. sterile solutions, isotonic solutions) are used. They are particularly administered by injection or infusion techniques.
  • solutions e.g. sterile solutions, isotonic solutions
  • sprays and solutions to be applied in drop form are particular formulations.
  • solutions to be applied in drop form, gels and ointments arc exemplified formulations.
  • the pharmaceutical compositions according to the invention can be administered such that the dose of the active compound is in the range customary for activators of TLR7.
  • the dose is dependent, for example, on the specific compound used, the species treated, age, body weight, general health, sex and diet of the subject treated, mode and time of administration, rate of excretion, severity of the disease to be treated and drug combination.
  • Cytokines that may be induced by the administration of compounds according to the invention generally include interferon (1FN) and/or tumor necrosis factor-ct (TNF-a) as well as certain interleukins (IL). Cytokines whose biosynthesis may be induced by compounds of the invention include IFN-a, TNF-a, IL-1 , IL-6, IL-10 and IL-12, and a variety of other cytokines. Among other effects, cytokines inhibit virus production and tumor cell growth, making the compounds useful in the treatment of tumors and viral diseases.
  • the compounds of the invention affect other aspects of the innate immune response. For example, natural killer cell activity may be stimulated, an effect that may be due to cytokine induction.
  • the compounds may also activate macrophages, which in turn stimulates secretion of nitric oxide and the production of additional cytokines. Further, the compounds may cause proliferation and differentiation of T- and/or B-lymphocytes.
  • the immune response modifying effects of the compounds make them useful in the treatment of a wide variety of conditions. Because of their ability to induce the production of cytokines such as IFN-a and/or TNF-a, and IL-12, the compounds are particularly useful in the treatment of viral diseases and tumors.
  • This immunomodulating activity suggests that compounds of the invention are useful in treating diseases such as, but not limited to, viral diseases including genital warts; common warts; plantar warts; Hepatitis B; Hepatitis C; Herpes Simplex Type I and Type II; molluscum contagiosum; HIV; CMV; VZV; intraepithelial neoplasias such as cervical intraepithelial neoplasia; human papillomavirus (HPV) and associated neoplasias; fungal diseases, e.g.
  • viral diseases including genital warts; common warts; plantar warts; Hepatitis B; Hepatitis C; Herpes Simplex Type I and Type II; molluscum contagiosum; HIV; CMV; VZV; intraepithelial neoplasias such as cervical intraepithelial neoplasia; human papillomavirus (HPV) and associated
  • Candida aspergillus, and cryptococcal meningitis
  • neoplastic diseases e.g., basal cell carcinoma, hairy cell leukemia, Kaposi's sarcoma, renal cell carcinoma, squamous cell carcinoma, myelogenous leukemia, multiple myeloma, melanoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, and other cancers
  • parasitic diseases e.g., basal cell carcinoma, hairy cell leukemia, Kaposi's sarcoma, renal cell carcinoma, squamous cell carcinoma, myelogenous leukemia, multiple myeloma, melanoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, and other cancers
  • parasitic diseases e.g.
  • the invention also provides a method of treating a viral infection in an animal comprising administering an effective amount of a compound of Formula (I) to the animal.
  • An amount effective to treat or inhibit a viral infection is an amount that will cause a reduction in one or more of the manifestations of viral infection, such as viral lesions, viral load, rate of virus production, and mortality as compared to untreated control animals.
  • the precise amount will vary according to factors known in the art but is expected to be a dose as indicated above with respect to the activation of TLR7, or a dose of about 100 ng/kg to about 50 mg/kg, particularly about 10 ⁇ g/kg to about 5 mg/kg.
  • An amount effective to treat a neoplastic condition is an amount that will cause a reduction in tumor size or in the number of tumor foci.
  • the precise amount will vary according to factors known in the art but is expected to be a dose as indicated above with respect to the activation of TLR7, or a dose of about 100 mg/kg to about 50 mg/kg, particularly about 10 mg/kg to about 5 mg kg.
  • the compounds according to the invention can be prepared, for example, as described as follows and according to the following specified reaction steps, or, particularly, in a manner as described by way of example in the following examples.
  • the compounds according to the invention are isolated and purified in a manner known per se, e.g. by distilling off the solvent in vacuo and recrystallizing the residue obtained from a suitable solvent or subjecting it to one of the customary purification methods, such as column chromatography on a suitable support material.
  • Salts of the compounds of Formula (I) according to the invention can be obtained by dissolving the free compound in a suitable solvent (for example a ketone such as acetone, methylethylketone or methylisobutylketone, an ether such as diethyl ether, tetrahydrofuran or dioxane, a chlorinated hydrocarbon such as methylene chloride or chloroform, or a low molecular weight aliphatic alcohol such as methanol, ethanol or isopropanol) which contains the desired acid or base, or to which the desired acid or base is then added.
  • a suitable solvent for example a ketone such as acetone, methylethylketone or methylisobutylketone, an ether such as diethyl ether, tetrahydrofuran or dioxane, a chlorinated hydrocarbon such as methylene chloride or chloroform, or a low molecular weight aliphatic alcohol
  • the acid or base can be employed in salt preparation, depending on whether a mono- or polybasic acid or base is concerned and depending on which salt is desired, in an equimolar quantitative ratio or one differing therefrom.
  • the salts are obtained by filtering, reprecipitating, precipitating with a non-solvent for the salt or by evaporating the solvent. Salts obtained can be converted into the free compounds which, in turn, can be converted into salts.
  • pharmaceutically unacceptable salts which can be obtained, for example, as process products in the manufacturing on an industrial scale, can be converted into pharmaceutically acceptable salts by processes known to the person skilled in the art.
  • the compounds of Formula (I) according to the invention can be converted into their N-oxides, for example, with the aid of hydrogen peroxide in methanol or with the aid of m-chloroperoxybenzoic acid in dichloromethane.
  • the person skilled in the art is familiar with the reaction conditions for carrying out the N-oxidation.
  • Pure diastereomers and pure enantiomers of the compounds and salts according to the invention that are present in the form of such stereoisomers can be obtained, e.g. by asymmetric synthesis, by usin chiral starting compounds in synthesis and by splitting up enantiomeric and diastereomeric mixtures obtained in synthesis.
  • Enantiomeric and diastereomeric mixtures can be split up into the pure enantiomers and pure diastereomers by methods known to a person skilled in the art. Particularly, diastereomeric mixtures are separated by crystallization, in particular fractional crystallization, or chromatography. Enantiomeric mixtures can be separated, e.g. by forming diastereomers with a chiral auxiliary agent, resolving the diastereomers obtained and removing the chiral auxiliary agent.
  • chiral auxiliary agents for example, chiral acids can be used to separate enantiomeric bases and chiral bases can be used to separate enantiomeric acids via formation of diastereomeric salts.
  • diastereomeric derivatives such as diastereomeric esters can be formed from enantiomeric mixtures of alcohols or enantiomeric mixtures of acids, respectively, using chiral acids or chiral alcohols, respectively, as chiral auxiliary agents.
  • diastereomeric complexes or diastereomeric clathrates may be used for separating enantiomeric mixtures.
  • enantiomeric mixtures can be split up using chiral separating columns in chromatography. Another suitable method for the isolation of enantiomers is the enzymatic separation.
  • Microwave chemistry was performed using a Biotage® Initiator Robot Sixty system. Unless specified otherwise, all TLC were performed on S1O2 plates (silica gel coated with fluorescent indicator F254) and all preparative reversed phase HPLC/MS purifications were performed on an XTerra RP18 5 ⁇ 19x150 mm column, using a gradient system 0.1 % HC0 2 H/H 2 0/15 ⁇ 95% CH 3 CN. Samples were loaded on the reverse phase column using DMSO/AcOH/H 2 0 1/1/1 or CH 3 CN/MeOH/DMF 80/15/5. Unless specified otherwise, all HC1, NaOH, KOH or Li OH solutions are aqueous.
  • the compounds were either characterized via proton NMR in oVdimethylsulfoxide or d-chloroform on a 300 MHz or 400 MHz NMR instrument (Bruker) and by mass spectroscopy, generally recorded via HPLC/MS in a fast gradient on C 18-material and using ESI (electrospray ionization) or APCI (atmospheric-pressure chemical ionization) mode.
  • ESI electrospray ionization
  • APCI atmospheric-pressure chemical ionization
  • the magnetic nuclear resonance spectral properties refer to the chemical shifts ( ⁇ ) expressed in parts per million (ppm).
  • the relative area of the shifts in the *H NMR spectrum corresponds to the number ofhydrogen atoms for a particular functional type in the molecule.
  • the nature of the shift, as regards multiplicity, is indicated as singlet (s), broad singlet (br s), doublet (d), triplet (t), quartet (q), quintuplet (quint.) and multiplet or massif (m).
  • Step 1 4-hydroxyquinoline (250 g, 1.72 mol) was dissolved in propionic acid (200 mL) and the mixture was stirred at 125°C. Nitric acid (158 mL, 3.79 mol, 2.2 eq) was then added dropwise while maintaining the temperature of the reaction at 125°C. After finishing the addition, the reaction mixture was stirred at 125°C for 60 min and then cooled down to room temperature. The resulting precipitate was filtered off and washed successively with ethanol, water and finally ethanol. The remaining solid was recrystallized from hot ethanol, cooled down, filtered off and dried under reduced pressure to give 252.3 g (77%) of 3-nitroquinolin-4-ol as a beige solid.
  • Step 2 POCh (661 mL, 7.21 mol, 18.3 eq) was heated at 60°C and 3-nitroquinolin-4- ol (75 g, 0.39 mol) was added in portions. The resulting suspension was then stirred at 120°C for 3 h, then cooled down to room temperature and the solvent was removed under reduced pressure. The crude residue was poured on a mixture of ice (1 kg) and CH2CI2 (500 mL). The aqueous layer was discarded and a solid crystallized from the organic phase during the evaporation. The resulting solid was filtered off and washed with CH2CI2 to give 40.4 g (50%) of the desired substance as a light beige solid. No further purification.
  • Step 2 A round-bottom flask was charged with a magnetic stir bar, 3-nitroquinolin-4- ol (41.82 g, 219.90 mmol), anhydrous CH2CI2 (1.05 L) and anhydrous DMF (8.51 mL, 109.95 mmol, 0.5 eq) to get a beige suspension. After addition of thionyl chloride (34.01 g, 285.87 mmol, 1.3 eq) the reaction mixture was reflux ed for 5 h. The progress of the reaction was monitored by TLC (petroleum ether/EtOAc 1:1) and by HPLC/MS. The reaction mixture was then cooled down to room temperature and used as such for the next step.
  • Step 1 4-Chloro-3-nitroquinoline (preparation A) (15 g, 71.91 mmol) was dissolved in CH 2 C1 2 (100 mL) and triethylamine (19.99 mL, 143.81 mmol, 2 eq) was added in portions at room temperature. 4- Amino- 1 -butanol (8.68 mL, 93.48 mmol, 1.3 eq) was added dropwise to the resulting solution (caution!
  • Step 2 To a solution of 4-((3-nitroquinolin-4-yl)amino)butan- 1 -ol (42.8 g, 163.80 mmol) and 4-dimethylaminopyridine (800 mg, 6.54 mmol, 0.04 eq) in chloroform (350 mL) triethylamine (34.2 mL, 245.70 mmol, 1.5 eq) was added dropwise at room temperature followed by addition of tert-butyldimethylsilyl chloride (32.1 g, 212.94 mmol, 1.3 eq) in portions. The resulting mixture was stirred overnight at room temperature. Reaction monitoring by HPLC/MS indicated a complete reaction.
  • the vessel was placed on a PARR shaker and pressurized to 50 psi (3.5 kg/cm 2 ) with hydrogen.
  • the reaction was monitored by TLC (CH 2 Cl 2 /MeOH 100:5) and was judged to be complete after one hour.
  • the catalyst was removed by filtration through a small pad of CELITE ® Hyflo Supercel. The filter cake was washed with toluene (3x100 mL) and the filtrates were combined. The volatiles were removed under reduced pressure to afford 31.65 g (96%) of the desired product as a dark oil.
  • Step 4 A round bottom flask was charged with a magnetic stir bar, N 4 -(4-((tert- butyldimethylsilyl)oxy)butyl)quinoline-3,4-diamine (8.99 g, 26.02 mmol), tri ethyl orthopropionate (9.17 g, 52.03 mmol, 2 eq) and toluene (76 mL). The reaction was heated at reflux to facilitate removal of the ethanol byproduct until TLC monitoring (benzene/methanol/acetone 1 :1 :8) and HPLC/MS indicated a complete conversion after 20 h.
  • Step 1 A round-bottom flask was charged with a magnetic stir bar, commercially available tert-butyl (4-((3-ammoquinolin-4-yl)amino)butyl)carbamate (40.00 g, 121.05 mmol), 2- ⁇ [(benzyloxy)carbonyl](ethyl)amino ⁇ acetic acid (Cbz-N-Et-Gly- OH, 34.46 g, 145.27 mmol, 1.2 eq), 2-(3H-[l ,2,3]triazolo[4,5-b]pyridin-3-yl)-l , 1 ,3,3- tetramethylisouronium hexafluorophosphate(V) (HATU, 55.23 g, 145.27 mmol, 1 .2 eq), triethylamine (36.75 g, 363.16 mmol, 3 eq), 4-dimethylaminopyridine
  • the reaction mixture was then concentrated by distillation of the toluene-acetic acid azeotrope at reduced pressure and the residue was suspended in saturated aqueous NaHCCb (200 mL) and filtered off.
  • the filtrate was extracted with chloroform (3x200 mL), the aqueous layer was adjusted to pH 10-1 1 by addition of 3M NaOH and extracted with chloroform (9x100 mL).
  • the filter cake and the aqueous layer were discarded.
  • the collected organic layers were washed successively with brine (400 mL) and dried over Na 2 S04, filtered off and concentrated under reduced pressure.
  • Step 1 A round bottom flask was charged with a magnetic stir bar, l-(4-((tert- butyldimethylsilyl)oxy)butyl)-2-cthyl-lH-imidazo[4,5-c]quinoline (see preparation B) (9.97 g, 25.99 mmol) and chloroform (130 mL). Solid 3 -chl orobcnzoperoxoic acid (4.93 g, 28.59 mmol, 1.1 eq) was added in portions to the solution over 15 minutes and the mixture was stirred at room temperature for 1 hour.
  • Step 2 A round bottom flask was charged with a magnetic stirrer bar, l-(4-((tert- butyldimethylsilyl)oxy)butyl)-2-ethyl-lH-imidazo[4,5-c]quinoline 5-oxide (9.64 g, 24.12 mmol), chloroform (100 mL) and ammonia solution (32%. 100 mL).
  • Step 3 A round bottom flask was charged with a magnetic stirrer bar, l-(4-((tert- butyldimethylsilyl)oxy)butyl)-2-ethyI-lH-imidazo[4,5-c]quinolin-4-amine (8.57 g, 21.50 mmol), tetrahydrofuran (90 mL), water (90 mL) and acetic acid (270 mL) and the resulting reddish solution was stirred at 60°C until TLC monitoring (benzene/methanol/acetone 1 :1 :8) and HPLC/MS indicated a complete conversion after 16 h.
  • the reaction mixture was then allowed to cool down to 0°C and adjusted to pH 8 with a dropwise addition of 10M NaOH ( ⁇ 333 mL).
  • the aqueous layer was extracted with a mixture of ethyl acetate and ethanol (5x200 mL).
  • the organic layers were combined, concentrated under reduced pressure and the resulting solid was extracted with DMF to get rid of sodium acetate salts.
  • the extract was concentrated in vacuo to afford 6.78 g (92 %) of 4-(4-amino-2-ethyl- 1 H-imidazo[4,5-c]quinolin- 1 - yl)butan-l -ol acetate as a beige-brown solid.
  • Step 4 Thionyl chloride (7.04 g, 59.20 mmol, 3 eq) was added to a suspension of 4- (4-amino-2-ethyl- 1 H-imidazo[4, 5-c]quinolin- 1 -yl)butan- 1 -ol acetate (6.78 g, 19.73 mmol) in dichloroethane (300 mL) and the yellow-beige mixture was stirred at room temperature overnight. HPLC MS monitoring showed a complete conversion. The mixture was cooled down to 0°C and methanol (25 mL) was slowly added.
  • Step 6 A pear-shaped flask was charged with a magnetic stir bar.
  • 2-ethyl- 1 -(4- ((tetrahydro-2H-pyran-4-yl)amino)butyl)-l H-imidazo[4,5-c]quinolin-4-amine 43 mg, 0.1 17 mmol; prepared by ion exchange chromatography on Agilent Stratospheres PL-HCO3 MP resin to remove traces of formic acid), 2M NaOH (88 ⁇ _, 0.176 mmol, 1 .5 eq) and water (1 mL) to give a pale yellow suspension.
  • R1 CH 2 N(Cbz)CH 2 CH 3
  • Step 1 A round-bottomed flask was charged with a magnetic stir bar, 4-(2-ethyl-lH- imidazo[4,5-c]quinolin-l -yl)butan-l -amine (see preparation D) (1.430 g, 5.329 mmol), tetrahydro -4H-pyran-4-one (533 mg, 5.329 mmol, 1 eq) and 1 ,2- dichloroethane (30 mL).
  • Step 2 A round bottom flask was charged with a magnetic stir bar, N-(4-(2-ethyl-lH- imidazo[4,5-c]quinolin- 1 -yl)butyl)tetrahydro-2H-pyran-4-amine (824 mg, 2.338 mmol), triethylamine (473 g, 4.675 mmol, 2 eq) and 1 ,2-dichloroethane (15 mL) to give a yellow solution. After addition of di-tert-butyl dicarbonate (510 mg, 2.338 mmol, 1 eq) the reaction mixture was stirred at room temperature overnight.
  • di-tert-butyl dicarbonate 510 mg, 2.338 mmol, 1 eq
  • Step 4 A round-bottom flask was charged with a magnetic stir bar, tert-butyl (4-(4- amino-2-ethyl- 1 H-imidazo[4,5-c]quinolin- 1 -yl)butyl)(tetrahydro-2H-pyran-4- yl)carbamate (602 mg, 1.287 mmol) and anhydrous 1 ,4-dioxane (10 mL) to give a yellow-beige suspension. 4N HCl/l ,4-dioxane (10 mL) was added and the reaction stirred at room temperature for 2.5 days.
  • Step 5 A round-bottom flask, was charged with a magnetic stir bar, 2-ethyl-l -(4- ((tetrahydro-2H-pyran-4-yl)amino)butyl)-lH-imidazo[4,5-c]quinolin-4- amine (367 mg, 0.999 mmol), triethylamine (101 mg, 0.999 mmol, 1 eq) and anhydrous chloroform (5 mL). The yellow solution was cooled to 0-4°C in an ice bath, trimethylsilyl isocyanate (127 mg, 1.099 mmol, 1.1 eq) was added and the mixture was stirred at room temperature.
  • Steps 1-5 Prepared as described in Example 2, using benzyl ((1 -(4-aminobutyl)-l H- imidazo[4,5-c]quinolin-2-yl)methyl)(ethy])carbamate (see preparation E) (3.5 g, 8.11 mmol) as starting material to yield 249 mg (5% overall yield for 5 steps) of benzyl ((4- amino- 1 -(4-(l -(tetrahydro-2H-pyran-4-yl)ureido)butyl)- 1 H-imidazo[4,5-c]quinolin-
  • Step 6 A round-bottom flask, equipped with a septum inlet flushing adapter with stopcock, was charged with a magnetic stir bar, benzyl ((4-amino- 1 -(4-( 1 -(tetrahydro- 2H-pyran-4-yl)ureido)butyl)-lH-imidazo[4,5-c]quinolin-2- yl )methyl)(ethyl)carbamate (245 mg, 0.427 mmol) and methanol (2 itiL) to give a pale yellow solution. After addition of 10% Pd/C (45 mg, 0.470 mmol) the apparatus was connected to a balloon filled with hydrogen and alternately evacuated and filled with hydrogen three times.
  • benzyl ((4-amino- 1 -(4-( 1 -(tetrahydro- 2H-pyran-4-yl)ureido)butyl)-lH-imidazo[4,5-c]quinolin-2- y
  • Step 1 A three-necked, 2-L round-bottomed flask was equipped with a 50 mm x 21 mm octagonal KOMETTM magnetic stir bar, a reflux condenser connected with a mineral oil bubbler and two glass stoppers. N-Boc-putrescine (56.41 g, 299.65 mmol, 1.5 eq), CH 2 C1 2 (500 mL) and molecular sieves 4A, powder ⁇ 5 micron (Aldrich) (90 g) were charged and the mixture was stirred at room temperature for 5 min.
  • N-Boc-putrescine 56.41 g, 299.65 mmol, 1.5 eq
  • CH 2 C1 2 500 mL
  • molecular sieves 4A powder ⁇ 5 micron (Aldrich)
  • the reaction mixture was then poured into cold water (1.4 L), the molecular sieves were removed by filtration and the dichioromethane layer was separated from the aqueous layer.
  • the aqueous layer was extracted with dichioromethane (3x250 mL) and the combined organic layers were washed with brine (2x250 mL), dried over Na 2 S04, filtered and concentrated under reduced pressure.
  • Step 2 A round-bottom flask was charged with a magnetic stir bar, a solution of tert- butyl (4-(N-(tetrahydro-2H-pyran-4-yl)acetamido)butyl)carbamate (62.81 g, 199.76 mmol) in dichloromethane (1.3 L). A solution of 4N HCl/1 ,4-dioxane (1.3 L) was carefully added and the resulting mixture was stirred at room temperature overnight. The progress of the reaction was monitored by TLC (CHC /MeOH/32% ammonia solution 240:9: 1 ) and the spots were visualized by treatment with ninhydrine reagent.
  • TLC CHC /MeOH/32% ammonia solution 240:9: 1
  • Step 3 A round-bottom flask was charged with a magnetic stir bar, N-(4-aminobutyl)- N-(tetrahydro-2H-pyran-4-yl)acetamide hydrochloride (crude, 51.46 g, 199.91 mmol), triethylamine (121.37 g, 1 199.43 mmol, 6 eq) and dichloromethane (2.1 L) and the resulting pale yellow solution was cooled to 0-4°C in an ice bath.
  • Step 4 A 250 mL PARR vessel (pressure vessel, Parr Instrument GmbH, Germany) was charged with N-(4-((3-nitroquinolin-4-yl)amino)butyl)-N-(tetrahydro-2H-pyran- 4-yl)acetamide (16.89 g, 43.71 mmol), 5% platinum on carbon (8.60 g, 2.19 mmoi, 5 mol%) and toluene (90 mL). The vessel was placed on a PARR shaker and pressurized to 50 psi (3.5 kg/cm 2 ). The reaction was monitored by TLC (CHCb/MeOH/32% ammonia solution 240:9: 1) and was complete after one hour.
  • TLC CHCb/MeOH/32% ammonia solution 240:9: 1
  • the catalyst was removed by filtration through a small pad of CELITE® Hyflo Super eel, the filter cake was washed several times with ethanol and the filtrates were combined. The volatiles were removed under reduced pressure and the crude substance was purified by flash chromatography on silica gel (CHCb/MeOH/32% ammonia solution 240:9: 1 to 140:9: 1 ) to afford 14.28 g (92%) of N-(4-((3-aminoquinolin-4-yl)amino)butyl)-N- (tetrahydro-2H-pyran-4-yl)acetamide as a red-orange oil.
  • Step 5 A round-bottom flask was charged with a magnetic stir bar, N-(4-((3- aminoquinolin-4-yl)amino)butyl)-N-(tetrahydro-2H-pyran-4-yl)acetamide (1.00 g, 2.81 mmol), N-Boc-N-ethylglycine (0.68 g, 3.37 mmol, 1.2 eq), 2-(3H- [ 1 ,2,3]triazolo[4,5-b]pyridin-3-yl)-l , 1 ,3,3-tetramethyIisouronium
  • hexafluorophosphate(V) (HATU, 1.28 g, 3.37 mmol, 1.2 eq), triethylamine (0.85 g, 8.42 mmol, 3 eq), 4-dimethylaminopyridine (0.03 g, 0.28 mmol, 0.1 eq) and anhydrous DMF (20 mL) to give a red-yellow solution.
  • the reaction mixture was stirred at room temperature until TLC monitoring (CHCb/MeOH/32% ammonia solution 140:9: 1 ) and HPLC/MS indicated complete conversion after about 10-12 hours. The solvent was then removed under reduced pressure and the residue, dissolved in ethyl acetate, was washed with water.
  • Step 6 A round bottom flask was charged with a magnetic stir bar, tert-butyl ethyl((I- (4-(N-(tetrahydro-2H-pyran-4-yl)acetamido)butyl)-lH-imidazo[4,5-c]quinolin-2- yl)mcthyl)carbamate (1.24 g, 2.37 mmol) and chloroform (100 mL).
  • Step 7 A round-bottom flask was charged with a magnetic stir bar, 2-(((tert- butoxycarbonyl)(ethyl)amino)methyl)- 1 -(4-(N-(tetrahydro-2H-pyran-4- yl)acetamido)butyl)-lH-imidazo[4,5-c]quinoline 5-oxide (1.33 g, 2.46 mmol), chloroform (30 mL) and ammonia solution (32%, 30 mL).
  • p-Toluenesulfonyl chloride (0.47 g, 2.46 mmol, 1 eq) was added to the biphasic mixture in one portion and the reaction mixture was vigorously stirred at room temperature overnight until TLC monitoring (CHCb/MeOH/32% ammonia solution 140:9:1 ) and HPLC/MS indicated complete conversion.
  • the reaction mixture was then diluted with chloroform (70 mL) and the phases were separated. The aqueous layer was adjusted to pH 7 with 2M HC1 and extracted with chloroform (2x100 mL). The combined organic layers were washed with brine (150 mL), dried over Na 2 S04, filtered and concentrated under reduced pressure.
  • Step 1 A round-bottom flask was charged with a magnetic stir bar, 4-(2-(2- methoxyethyl)-l H-imidazo[4,5-c]quinolin-l-yl)butan-l-ol (see preparation C) (9.48 g crude material, corresponding to -14.47 mmol starting material ) and dichloroethane (400 mL) to give a yellow-brown solution. After addition of thionyl chloride (6.89 mL, 95.00 mmol, 6.5 eq), the mixture was stirred at room temperature for 12 h until TLC monitoring (CH2Cl 2 /MeOH 9: 1) and HPLC/MS indicated complete reaction.
  • Step 2 A reaction flask was charged with a magnetic stir bar, 1 -(4-chlorobutyl)-2-(2- methoxyethyl)- 1 H-imidazo[4,5-c]quinoline hydrochloride (3.35 g, 9.46 mmol) and water (300 mL) to give a reddish-brown suspension. After addition of magnesium monoperoxyphthalate hexahydrate (MMPP) (4.68 g, 9.46 mmol, 1 eq) the mixture was stirred at 60°C.
  • MMPP magnesium monoperoxyphthalate hexahydrate
  • magnesium monoperoxyphthalate hexahydrate (4.68 g, 9.46 mmol, 1 eq) was added and the mixture was further stirred at 60°C for 90 min until TLC monitoring (CH 2 Cl 2 MeOH 9:1) and HPLC/MS indicated complete reaction.
  • the reaction mixture was extracted with chloroform (3x100 mL) and the combined organic layers were washed with saturated sodium bicarbonate solution (1x50 mL).
  • Step 5 A pear-shaped flask was charged with a magnetic stir bar, 2-(2-methoxyethyl)- 1 -(4-((tetrahydro-2H-pyran-4-yl)amino)butyl)- 1 H-imidazo[4,5-c]quinolin-4-amine (302 mg, 0.760 mmol), 2N NaOH solution (3.799 mL, 7.597 mmol, 10 eq) and water (5 mL) to give a yellow solution. After addition of acetic anhydride (0.776 g, 7.597 mmol, 10 eq), the reaction mixture was stirred at room temperature for 24 h and the progress of the reaction was monitored by HPLC/MS.
  • Step 1 4-(2-Ethyl- 1 H-imidazof 4,5-c]quinolin- 1 -yObutan- 1 -amine (see preparation D) (10.4 g, 38.7 mmol) was suspended in MeOH/THF (1 :1.5 v/v, 150 mL) and a solution oftriethylamine (51.9 mL, 373.5 mmol, 9.65 eq) in MeOH/THF (1 : 1 v/v, 20 mL) was added dropwise.
  • Step 2 To a solution of 3-((4-(2-ethyl-l H-imidazo[4,5-c]quinolin-l - yl)butyl)amino)thietane 1,1 -dioxide (3.14 g, 8.42 mmol) in CH2CI2 (30 mL) at 0°C were added triethylamine (3.5 mL, 25.3 mmol, 3 eq) dissolved in CH 2 C1 2 (5 mL) and acetic acid anhydride (3.1 mL, 33.7 mmol, 4 eq) dissolved in CH2CI2 (5 mL).
  • Step 3 To a magnetically stirred solution of N-(l ,l-dioxidothietan-3-yl)-N-(4-(2- ethyl- 1 H-imidazo[4,5-c]quinolin- 1 -yl)butyl)acetamide (330 mg, 0.796 mmol) in CH 2 Ck (20 mL) was added peroxyacetic acid (38-40% in acetic acid, 398 ⁇ _, 2.388 mmol, 3 eq) at room temperature and the resulting mixture was then stirred at reflux for 3 h. The completion of the conversion was monitored by HPLC/MS analysis. The mixture was then cooled down to room temperature, diluted with water (40 mL) and stirred for 5 min.
  • dioxidothietan-3-yl)urea Prepared as described in Example 7, starting with 80 mg (0.215 mmol) of 3-((4-(2- ethyl-l H-imidazo[4,5-c]quinolin-l -yl)butyl)amino)thietane 1 ,1 -dioxide (from step 1) and using trimethylsilyl isocyanate (4 eq) for the formation of the urea to yield 8.5 mg (9% overall yield for 3 steps) of a beige solid.
  • Example 10 (for illustrative purposes)
  • Steps 1-3 Prepared as described in Example 7, using benzyl ((1 -(4-aminobutyl)- 1 H- imidazo[4,5-c]quinolin-2-yl)methyl)(ethyl)carbamate (see preparation E) (1 g, 2.317 mmol) as starting material to yield 261 mg (19% over 3 steps) of benzyl ((4-amino- 1 - (4-(N-(l , 1 -dioxidothietan-3-yl)acetamido)butyl)- lH-imidazo[4,5-c]quinolin-2- yl)methyl)(ethyl)carbamate as an off-white foam.
  • Step 4 A round-bottom flask, equipped with a septum inlet flushing adapter with stopcock, was charged with a magnetic stir bar, benzyl ((4-amino- l-(4-(N-( 1,1- dioxidothietan-3 -yl)acetamido)butyl)- 1 H-imidazo[4,5-c]quinolin-2- yl )rnethyl)(ethyl)carbamate (261 mg, 0.440 mmol) and methanol (20 mL) to give a colorless solution.
  • benzyl ((4-amino- l-(4-(N-( 1,1- dioxidothietan-3 -yl)acetamido)butyl)- 1 H-imidazo[4,5-c]quinolin-2- yl )rnethyl)(ethyl)carbamate (261 mg, 0.440 mmol) and methanol (20 mL)
  • the filter cake was washed with methanol (3x50 mL).
  • the combined filtrates were filtered through a 0.45 ⁇ PTFE membrane to remove catalyst residues and concentrated under reduced pressure.
  • the residue was purified by preparative TLC (Si0 2 20 cm 2 , CHCb/MeOH/32% ammonia solution 90:9:1) to give 108 mg (53%) of an off-white solid.
  • Example 11 (for illustrative purposes)
  • Example 12 (for illustrative purposes)
  • Step 1 A 50 mL two-neck round-bottom flask, equipped with a reflux condenser, was charged with a magnetic stir bar, benzyl (( 1 -(4-aminobutyl)- 1 H-imidazo[4,5- c]quinolin-2-yl)methyl)(ethyl)carbamate (see preparation E) (1.000 g, 2.317 mmol), 1 , 1 -dioxidothietan-3-yl methanesul fonate (557 mg, 2.781 mmol, 1.2 eq), DIPEA (1.797 g, 13.904 mmol, 6 eq) and a mixture of THF/water (4: 1 v/v, 20 mL) to give a yellow suspension that was heated under reflux until TLC monitoring (CHCh/MeOH/32% ammonia solution 90:9: 1) and HPLC/MS indicated a complete consumption of the starting material after 2 h.
  • TLC monitoring CHCh/MeOH
  • Step 2 A round bottom flask was charged with a magnetic stir bar, crude tert-butyl (4- (2-((((benzyloxy)carbonyl)(ethyl)amino)methyl)-lH-imidazo[4,5-c]quinolin-l- yl)butyl)(l , 1 -dioxidothietan-3-yl)carbamate (1.480 g, 2.328 mmol) and CH 2 C1 2 (20 mL) to give a reddish-yellow solution.
  • Triethylamine (283 mg, 2.793 mmol, 1.2 eq), 4-dimethylaminopyridine (28 mg, 0.233 mmol, 0.1 eq) and benzyl chloroformate (477 mg, 2.793 mmol, 1.2 eq) were then added and the reaction mixture was stirred at room temperature for 2 h.
  • Step 4 A 10-mL round-bottom flask equipped with a magnetic stir bar was charged with benzyl ((4-(((benzyloxy)carbonyl)amino)- 1 -(4-((tert-butoxycarbonyl)( 1 , 1 - dioxidothietan-3-yl)amino)butyl)- 1 H-imidazo[4,5-c]quinolin-2- yl)methyl)(ethyl)carbamate (257 mg, 0.327 mmol) and anhydrous 1 ,4-dioxane (5 mL) to give a yellow solution.
  • Step 5 A round-bottom flask was charged with a magnetic stir bar, benzyl ((4- (((benzyloxy)carbonyl)amino)- 1 -(4-(( 1 , 1 -dioxidothietan-3-yl)amino)butyl)- 1 H- imidazo[4,5-c]quinolin-2-yl)methyl)(ethyl)carbamate (230 mg, 0.336 mmol), triethylamine (47 ⁇ L, 0.336 mmol, 1 eq) and anhydrous chloroform (5 mL) and the resulting yellow solution was cooled to 0-4°C in an ice bath.
  • benzyl ((4- (((benzyloxy)carbonyl)amino)- 1 -(4-(( 1 , 1 -dioxidothietan-3-yl)amino)butyl)- 1 H- imidazo[4,5-c]quinolin
  • Step 6 A round-bottom flask, equipped with a septum inlet flushing adapter with stopcock, was charged with a magnetic stir bar, benzyl ((4- (((benzyloxy)carbonyl)amino)- 1 -(4-( 1-(1,1 -dioxidothietan-3 -yl)ureido)butyl)- 1 H- imidazo[4,5-c]quinolin-2-yl)methyl)(ethyl)carbamate (97 mg, 0.133 mmol) and methanol (20 mL) to give a colorless suspension.
  • benzyl ((4- (((benzyloxy)carbonyl)amino)- 1 -(4-( 1-(1,1 -dioxidothietan-3 -yl)ureido)butyl)- 1 H- imidazo[4,5-c]quinolin-2-yl)methyl)(ethyl)carbamate (97 mg, 0.133
  • Step 1 To a solution of 1 -(4-chlorobutyl)-2-(2-methoxyethyl)- 1 H-imidazo[4,5- c]quinolin-4-amine (obtained in Example 5, Steps 1-3) (286 mg, 0.859 mmol) in anhydrous DMA (10 mL) were added sodium iodide (129 mg, 0.859 mmol, 1 eq), N- ethyl-N-isopropylpropan-2-amine (449 ⁇ _, 2.578 mmol, 3 eq), thietan-3 -amine hydrochloride (341 mg, 2.578 mmol, 3 eq) and molecular sieve 4A, powder ⁇ 5 micron (Aldrich) (1.5 g).
  • sodium iodide 129 mg, 0.859 mmol, 1 eq
  • N- ethyl-N-isopropylpropan-2-amine (449 ⁇ _, 2.578
  • the yellow reaction mixture was stirred at 100°C until HPLC/MS monitoring indicated complete reaction and formation of the desired substance after 4 days.
  • the reaction mixture was filtered off and concentrated under reduced pressure to get 504 mg (crude) of 2-(2-methoxyethyl)- 1 -(4-(thietan-3 -ylamino)butyl)- 1 H- imidazo[4,5-c]quinolin-4-amine as a brown residue.
  • the raw substance was used for the next step without any further purification.
  • Step 2 A pear-shaped flask was charged with a magnetic stir bar, crude 2-(2- methoxyethyl)- 1 -(4-(thietan-3 -ylamino)butyl)- 1 H-imidazo[4,5-c]quinolin-4-amine (504 mg, 1.3 mmol), 2M NaOH (6.536 mL, 13.07 mmol, 10 eq) and water (5 mL) to give a brownish suspension. After addition of acetic anhydride (1.22 mL, 13.07 mmol, 10 eq), the reaction mixture was stirred at room temperature overnight. HPLC/MS monitoring indicated complete consumption of the starting material.
  • Step 3 A pear-shaped flask was charged with a magnetic stir bar, N-(4-(4-amino-2- (2-methoxyethyl)- 1 H-imidazo[4,5-c]quinolin- 1 -yl)butyl)- -(thietan-3 -yl)acetamide (91 mg, 0.213 mmol), chloroform (5 mL) and methanol (5mL) to give a light yellow solution. Solid 3-chlorobenzoperoxoic acid (1 10 mg, 0.638 mmol, 3 eq) was added in portions to the solution and the reaction was stirred at room temperature overnight. HPLC/MS monitoring indicated an incomplete reaction.
  • Step 2 Methyl chloroformate (21 ⁇ , 0.27 mmol, 1 eq) was added at room temperature to a suspension of 3-((4-(4-amino-2-(2-methoxyethyl)-l H-imidazo[4,5-c]quinolin- 1 - yl)butyl)amino)thietane 1 ,1 -dioxide hydrochloride (114 mg, 0.27 mmol) and K 2 C0 3 (94 mg, 0.81 mmol, 3 eq) in H 2 0 (3 mL) and the mixture was stirred overnight at room temperature. HPLC/MS monitoring indicated an incomplete conversion.
  • the reaction was monitored by TLC (CH 2 Cl 2 /CHC1.3/methanol/32% ammonia solution 100:80:18:2) and HPLC/MS. Additional trimethylsilyl isocyanate (192 ⁇ _, 1.40 mmol, 2.2 eq) and further stirring at room temperature overnight were necessary to obtain a complete consumption of the starting material.
  • the reaction mixture was then filtered off and the filtrate was diluted with CH 2 C1 2 (200 mL) and washed with saturated aqueous NaHC0 3 .
  • the aqueous phase was then extracted twice with CH 2 C1 2 and the combined organic layers were dried over Na 2 S0 4 , filtered off and concentrated under reduced pressure.
  • the crude substance was purified by preparative TLC (CH 2 Cl 2 /methanol/32% ammonia solution 150:9:1 then 180:18:2) to yield 34 mg (11 %) of an off-white solid.
  • BSA bovine serum albumine
  • PBS phosphate buffer pH 7.4
  • PE phycoerythrin
  • EDC l-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride.
  • PBMCs were pre-incubated in medium (RPMI1640 -Gibco #61870-044 + 10% FBS + 1% L-glutamine) for 5 hours at 37°C and 5% CO2 before the compounds of the present invention or controls were added, respectively.
  • medium RPMI1640 -Gibco #61870-044 + 10% FBS + 1% L-glutamine
  • the cells are incubated for another 24h at 37°C and 5% C0 2 .
  • Luminex assay Step 1 : Coupling of antibody to beads:
  • TLR stimulation assay Compounds are tested for stimulation of human TLR7 and TLR8, respectively, via reporter gene expression analyzed in a cell line stably transfected with human TLR7 and TLR8, respectively, and coexpressing an NFkB/AP- inducible SEAP reporter gene. Stimulation with a TLR ligand leads to NFkB/AP-1 activation and upregulation and secretion of SEAP, which can be measured in the supernatant with QuantiBlue detection system (InvivoGen).
  • HEK-Blue-hTLR7 InvivoGen #hkb-htlr7
  • HE -Blue- hTLR8 InvivoGen #hkb-htlr8
  • QuantiBlueTM InvivoGen #10H19-MM solution, following manufacturer's instructions: solubilize the powder of one pouch in 100ml MilliQ grade water, heat at 37°C in a water bath for about 30 min, filter the solution with a paper filter
  • the compound of Example 1 showed an EC50 of 44 nM in the TLR7 reporter gene assay.
  • 3- ⁇ acetyl[4-(4-amino-2-ethyl- 1 H-imidazo[4,5-c]quinolin- 1 - yl)butyl] amino ⁇ -2,5 -anhydro- 1 , 3 ,4-trideoxypentitoI a comparative compound described in WO-A-2009/U 8296 as example III, showed a ten-fold higher EC 5 o of 440 nM in the TLR7 reporter gene assay.
  • the compound of Example 1 showed no activity towards TLR8 in the above described TLR8 reporter gene assay.
  • the inventors of the present application have found that the double substitution of the amino group NR2R3 of Formula (I) is associated with the TLR7 selectivity, whereas a mono-substitution of the amino group NR2R3 of Formula (I) (i.e., where R2 is H) leads to compounds which are selective for TLR8.
  • Example - In vivo profiling In the following studies, compounds according to the invention were examined in an in vivo cynomolgus monkey model. It is evaluated whether and at which applied dose the compounds cause secretion of IFN-ct.
  • test compound was applied intravenously to cynomolgus monkeys by a thirty minute infusion.
  • blood samples 0.5 mL for approximately 0.25 mL plasma
  • Plasma was obtained by centrifugation at 4°C and approximately 1800 g for 10 minutes and was aliquoted into labeled micro tubes and stored frozen at-70°C or below.
  • Plasma samples were thawed, diluted and used for determination of IFN-a levels using an IFN-a Elisa Kit (e.g. VeriKineTM Cynomolgus/Rhesus IFN-a ELISA Kit) according to the manufacturer's instructions. Results show that compounds of the present invention cause IFN-a secretion in vivo in cynomolgus monkeys, whereas application of vehicle does not result in measurable IFN-a levels. Blood plasma peak concentrations of IFN-a are usually reached approximately 150 minutes after initiation of compound application.
  • IFN-a Elisa Kit e.g. VeriKineTM Cynomolgus/Rhesus IFN-a ELISA Kit

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

L'invention concerne des dérivés d'imidazoquinoline et des compositions pharmaceutiques contenant les dérivés d'imidazoquinoline. Les dérivés d'imidazoquinoline de l'invention sont utiles en tant qu'agonistes du récepteur de type Toll, en particulier des agonistes de TLR7, et favorisent l'induction de certaines cytokines.
EP18759137.5A 2017-09-06 2018-08-31 Imidazoquinolines substituées Active EP3679038B1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PCT/EP2017/072352 WO2019048036A1 (fr) 2017-09-06 2017-09-06 Imidazoquinolines substituées
PCT/EP2018/073470 WO2019048350A1 (fr) 2017-09-06 2018-08-31 Imidazoquinolines substituées

Publications (2)

Publication Number Publication Date
EP3679038A1 true EP3679038A1 (fr) 2020-07-15
EP3679038B1 EP3679038B1 (fr) 2024-04-03

Family

ID=59914433

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18759137.5A Active EP3679038B1 (fr) 2017-09-06 2018-08-31 Imidazoquinolines substituées

Country Status (16)

Country Link
US (2) US11267813B2 (fr)
EP (1) EP3679038B1 (fr)
JP (1) JP7241738B2 (fr)
KR (1) KR20200050963A (fr)
CN (1) CN111094287A (fr)
AU (1) AU2018327414B2 (fr)
BR (1) BR112020004504A2 (fr)
CA (1) CA3074166A1 (fr)
DK (1) DK3679038T3 (fr)
FI (1) FI3679038T3 (fr)
IL (1) IL273002B2 (fr)
MX (1) MX2020002367A (fr)
RU (1) RU2768629C2 (fr)
SG (1) SG11202001925WA (fr)
WO (2) WO2019048036A1 (fr)
ZA (2) ZA202001266B (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019048036A1 (fr) * 2017-09-06 2019-03-14 Biontech Ag Imidazoquinolines substituées
LT3679025T (lt) 2017-09-06 2022-06-27 BioNTech SE Pakeistieji imidazochinolinai, kaip tlr7 agonistai
SG11202108752YA (en) * 2019-03-07 2021-09-29 BioNTech SE Process for the preparation of a substituted imidazoquinoline
WO2021086689A1 (fr) * 2019-10-29 2021-05-06 Prime Reach Trading Limited Composés de 4-amino-imidazoquinoline et utilisation de ceux-ci
WO2023083439A1 (fr) * 2021-11-09 2023-05-19 BioNTech SE Agoniste de tlr7 et combinaisons pour le traitement du cancer

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS585623B2 (ja) 1976-07-12 1983-02-01 日本電信電話株式会社 ファクシミリ通信方式
US6573273B1 (en) 1999-06-10 2003-06-03 3M Innovative Properties Company Urea substituted imidazoquinolines
US6756382B2 (en) 1999-06-10 2004-06-29 3M Innovative Properties Company Amide substituted imidazoquinolines
US6451810B1 (en) 1999-06-10 2002-09-17 3M Innovative Properties Company Amide substituted imidazoquinolines
US6677349B1 (en) 2001-12-21 2004-01-13 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US7223738B2 (en) 2002-04-08 2007-05-29 Merck & Co., Inc. Inhibitors of Akt activity
CA2547085A1 (fr) 2003-11-25 2005-06-09 3M Innovative Properties Company Hydroxylamine, et imidazoquinoleines, et imidazopyridines et imidazonaphtyridine substitues d'oxime
JP5128940B2 (ja) * 2004-06-18 2013-01-23 スリーエム イノベイティブ プロパティズ カンパニー 置換イミダゾキノリン、イミダゾピリジン、およびイミダゾナフチリジン
KR20080083270A (ko) * 2005-11-04 2008-09-17 콜레이 파마시티컬 그룹, 인코포레이티드 하이드록시 및 알콕시 치환된 1에이치 이미다조퀴놀린 및방법
US9370508B2 (en) 2007-02-20 2016-06-21 Novartis Ag Imidazoquinolines as dual lipid kinase and mTOR inhibitors
US8436178B2 (en) * 2007-05-08 2013-05-07 Astrazeneca Ab Imidazoquinolines with immuno-modulating properties
NZ588183A (en) 2008-03-24 2012-05-25 4Sc Discovery Gmbh Novel substituted imidazoquinolines
EA022629B1 (ru) 2009-04-16 2016-02-29 Фундасьон Сентро Насиональ Де Инвестигасьонес Онколохикас Карлос Iii Имидазопираны для применения в качестве ингибиторов киназ
CN103012398B (zh) 2011-09-19 2015-10-14 上海恒瑞医药有限公司 咪唑并喹啉类衍生物及其可药用盐、其制备方法及其在医药上的应用
WO2019048036A1 (fr) * 2017-09-06 2019-03-14 Biontech Ag Imidazoquinolines substituées
LT3679025T (lt) 2017-09-06 2022-06-27 BioNTech SE Pakeistieji imidazochinolinai, kaip tlr7 agonistai

Also Published As

Publication number Publication date
EP3679038B1 (fr) 2024-04-03
WO2019048350A1 (fr) 2019-03-14
IL273002B1 (en) 2023-04-01
FI3679038T3 (fi) 2024-05-23
RU2768629C2 (ru) 2022-03-24
CA3074166A1 (fr) 2019-03-14
KR20200050963A (ko) 2020-05-12
JP2020532570A (ja) 2020-11-12
CN111094287A (zh) 2020-05-01
RU2020112229A3 (fr) 2021-10-06
AU2018327414B2 (en) 2024-02-15
SG11202001925WA (en) 2020-04-29
ZA202001266B (en) 2021-08-25
MX2020002367A (es) 2020-07-20
AU2018327414A1 (en) 2020-03-19
US11267813B2 (en) 2022-03-08
US20220227756A1 (en) 2022-07-21
US20200317663A1 (en) 2020-10-08
BR112020004504A2 (pt) 2020-09-15
IL273002B2 (en) 2023-08-01
DK3679038T3 (da) 2024-05-27
ZA202101322B (en) 2022-09-28
RU2020112229A (ru) 2021-10-06
JP7241738B2 (ja) 2023-03-17
WO2019048036A1 (fr) 2019-03-14
IL273002A (en) 2020-04-30

Similar Documents

Publication Publication Date Title
AU2018327414B2 (en) Substituted imidazoquinolines
US11987569B2 (en) Substituted imidazoquinolines as agonists of TLR7
ES2359123T3 (es) Imidazopiridinonas.
JP2019070042A (ja) インドールアミン2,3−ジオキシゲナーゼの阻害剤としての活性を有するベンゾイミダゾールおよびイミダゾピリジンカルボキシミドアミド化合物
JP5830094B2 (ja) 置換イミダゾ[1,2−a]ピリミジンおよび−ピリジン
CN113272305A (zh) 吲哚ahr抑制剂及其用途
WO2013071698A1 (fr) Inhibiteur tricyclique de pi3k et/ou mtor
CA3159842A1 (fr) Medicament conjugue de polyethylene glycol, son procede de preparation et son utilisation
US20220227762A1 (en) Amide-substituted imidazo compounds as selective inhibitors of indoleamine 2,3-dioxygenases
RU2776748C2 (ru) Замещенные имидазохинолины в качестве агонистов tlr7

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20200316

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40022363

Country of ref document: HK

DAV Request for validation of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20210419

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20230614

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTC Intention to grant announced (deleted)
INTG Intention to grant announced

Effective date: 20231026

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602018067520

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20240524

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20240425